EP2626073A1 - Verbindung zur Verwendung zur Prävention bzw. Behandlung einer neurogenerativen Erkrankung oder einer Erkrankung mit Phosphodiesterase (PDE4) -4-Aktivierung - Google Patents

Verbindung zur Verwendung zur Prävention bzw. Behandlung einer neurogenerativen Erkrankung oder einer Erkrankung mit Phosphodiesterase (PDE4) -4-Aktivierung Download PDF

Info

Publication number
EP2626073A1
EP2626073A1 EP12181152.5A EP12181152A EP2626073A1 EP 2626073 A1 EP2626073 A1 EP 2626073A1 EP 12181152 A EP12181152 A EP 12181152A EP 2626073 A1 EP2626073 A1 EP 2626073A1
Authority
EP
European Patent Office
Prior art keywords
alkyl
represent
hydrogen atom
compound
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP12181152.5A
Other languages
English (en)
French (fr)
Inventor
Michel Souchet
Arnaud Sinan Karaboga
Thierry Pillot
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Harmonic Pharma
Synaging
Original Assignee
Harmonic Pharma
Synaging
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Harmonic Pharma, Synaging filed Critical Harmonic Pharma
Priority to PCT/EP2013/052867 priority Critical patent/WO2013120896A1/en
Publication of EP2626073A1 publication Critical patent/EP2626073A1/de
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the invention relates to the field of the prevention and/or treatment of diseases involving an activation of phosphodiesterase-4 (PDE4).
  • the present invention relates to modulators of phosphodiesterase-4 (PDE4), preferably allosteric modulators of PDE4, for use in the prevention and/or treatment of a disease involving an activation of PDE4, such as for example a neurodegenerative disease.
  • said modulators of phosphodiesterase-4 have the following general formula (I): or a pharmaceutically acceptable salt or solvate thereof, wherein X and Z each independently represent CH or N; Y represents O or S; R1, R2 and R3 may be the same or different and represent a hydrogen atom, a halogen atom, hydroxyl, nitro, cyano, amino, amido, carbamate, alkyl, alkenyl, alkynyl, alkoxy, heterocyclic, carbocyclic, alkylthio, carboxy, or alkoxycarbonyl; R4 represent a hydrogen atom; R5, R6, R7 and R8 may be the same or different and represent a hydrogen atom, halogen atom, hydroxyl, nitro, amino, alkyl, alkoxy, alkylthio, or trifluoromethyl; R9 and R10 may be the same or different and represent a hydrogen atom, alkyl, alkylcarbonyl, heterocyclic
  • the disease involving an activation of PDE4 is a neurodegenerative disease such as for example Alzheimer disease, Parkinson disease, multiple sclerosis or amyotrophic lateral sclerosis.
  • Phosphodiesterase-4 is an enzyme belonging to the phosphodiesterase family. Phosphodiesterases are enzymes that break phosphodiester bonds. Especially, phosphodiesterase-4 (PDE4) regulates the intracellular level of cyclic adenosine (cAMP) by hydrolyzing cAMP within both immune cells and cells in the central nervous system (CNS). PDE4 inhibitors prevent the inactivation of the intracellular messenger cAMP.
  • cAMP cyclic adenosine
  • PDE4 is therefore a therapeutic target of high interest for neurological diseases, neurodegenerative diseases, inflammatory diseases, respiratory diseases, allergies, cardiovascular diseases, thrombosis, diabetes, cancer, rheumatoid arthritis, atopic dermatitis, psoriasis, asthma, inflammatory bowel disease.
  • Neurodegenerative diseases are characterized by a progressive and irreversible deterioration of brain cells and eventually death of the neurons.
  • Neurodegenerative diseases referred to in the present invention may or may not be associated with neuroinflammatory processes or to PDE4 activation processes.
  • Neurodegenerative diseases comprise among others, but are not limited to, Alzheimer disease, Parkinson disease, multiple sclerosis or amyotrophic lateral sclerosis, and any disease wherein a progressive loss of structure or function of neurons, including death of neurons, occurs.
  • Efficient treatments for preventing, stopping and/or reversing disease involving an activation of PDE4, especially neurodegenerative diseases are needed.
  • PDE4 inhibitors have yet been brought to market because of issues related to tolerability and side effects such as emesis and diarrhea.
  • the emetic response to PDE4 inhibitors is mediated in part by a brainstem noradrenergic pathway and can be reduced by limiting distribution to the brain.
  • CNS central nervous system
  • PDE4 inhibitors that have been explored in human clinical trials bind the active site competitively with cAMP and therefore completely inhibit enzyme activity at high concentrations. Although this traditional approach to PDE4 inhibitor design has demonstrated therapeutic benefit, competitive inhibitors are likely to alter cAMP concentrations beyond normal physiological levels, perturbing the tight temporal and spatial control of cAMP signaling within cells, and leading to side effects.
  • PDE4 allosteric modulators that only partially inhibit cAMP hydrolysis. Such compounds are more likely to lower the magnitude of PDE4 inhibition and maintain cAMP signaling, thereby reducing target-based toxicity.
  • a similar approach has been described previously for allosteric modulators of G protein-coupled receptors and for atypical retinoid ligands of nuclear hormone receptors.
  • PDE4 allosteric modulators are potent in cellular and in vivo assays and have greatly reduced potential for emesis.
  • PDE4 allosteric modulators for the treatment of diseases involving an activation of PDE4 of the invention was performed by a unique in silico selection process developed by the Applicant. The activity of in silico selected compounds was then confirmed by in vitro and in vivo assays.
  • the Applicant used its proprietary method of generating spherical harmonic (SH) based molecular representation of compounds of therapeutic interest.
  • SH spherical harmonic
  • small molecules may be converted into their spherical harmonic (SH) based molecular representation by successive deflation processes.
  • This method was applied to known PDE4 allosteric modulators to determine a common SH shape.
  • the cognate SH shapes of compound D159687 was calculated as represented on figure 2 .
  • Compound D159687 was described as being a PDE4 allosteric modulators by Burgin et al. in Nature Biotechnology, Vol. 28, Number 1, January 2010, pp. 63-72 .
  • the present invention thus relates to compounds of general formula (I) or a pharmaceutically acceptable salt or solvate of thereof, for use in the prevention and/or treatment of a neurodegenerative disease and/or a disease involving an activation of PDE4, said formula (I) being the following: wherein X and Z each independently represent CH or N; Y represents O or S; R1, R2 and R3 may be the same or different and represent a hydrogen atom, a halogen atom, hydroxyl, nitro, cyano, amino, amido, carbamate, alkyl, alkenyl, alkynyl, alkoxy, heterocyclic, carbocyclic, alkylthio, carboxy, or alkoxycarbonyl; R4 represent a hydrogen atom; R5, R6, R7 and R8 may be the same or different and represent a hydrogen atom, halogen atom, hydroxyl, nitro, amino, alkyl, alkoxy, alkylthio, or trifluoromethyl; R9 and R
  • the present invention relates to tivozanib or a pharmaceutically acceptable salt or solvate of thereof, for use in the prevention and/or treatment of a disease selected from a neurodegerative disease and a disease involving an activation of PDE4, preferably the treatment of Alzheimer disease, wherein so-called compound tizovanib (Av-951, KRN951), is of formula:
  • the present invention relates to 1-(2-chloro-4-((6,7-dimethoxyquinazolin-4-yl)oxy)phenyl)-3-propylurea or a pharmaceutically acceptable salt or solvate of thereof, for use in the prevention and/or treatment of a disease involving an activation of PDE4, of following formula:
  • substituents may be selected from but not limited to, for example, the group comprising halogen, hydroxyl, oxo, nitro, amido, carboxy, amino, cyano, haloalkoxy, haloalkyl, alkyl 1, alkenyl, alkynyl, carbocyclic group, heterocyclic group, carbocyclic group, azolyl, alkoxy, alkoxycarbonyl, alkylcarbonyl, alkylthio, trifluoromethyl, heterocycliccarbonyl, carbocycliccarbonyl.
  • This invention also relates to a compound of formula (I) below for use in the prevention and/or the treatment of a neurodegenerative disease, such as for example Alzheimer disease, Parkinson disease, multiple sclerosis or amyotrophic lateral sclerosis, especially Alzheimer disease or for the prevention and/or the treatment of a disease involving an activation of PDE4.
  • a neurodegenerative disease such as for example Alzheimer disease, Parkinson disease, multiple sclerosis or amyotrophic lateral sclerosis, especially Alzheimer disease or for the prevention and/or the treatment of a disease involving an activation of PDE4.
  • the compound of the invention for use for preventing and/or treating a disease involving an activation of PDE4 is a modulator of PDE4, preferably an allosteric modulator of PDE4.
  • the compound for use in the prevention and/or treatment of a neurodegenerative disease or a disease involving an activation of phosphodiesterase-4 is a compound of general formula (I) below or a pharmaceutically acceptable salt or solvate thereof: wherein X and Z each independently represent CH or N; Y represents O or S; R1, R2 and R3 may be the same or different and represent a hydrogen atom, a halogen atom, hydroxyl, nitro, cyano, amino, amido, carbamate, alkyl, alkenyl, alkynyl, alkoxy, heterocyclic, carbocyclic, alkylthio, carboxy, or alkoxycarbonyl; R4 represent a hydrogen atom; R5, R6, R7 and R8 may be the same or different and represent a hydrogen atom, halogen atom, hydroxyl, nitro, amino, alkyl, alkoxy, alkylthio, or trifluoromethyl; R9 and R10 may be the same
  • X represents CH. In another embodiment, X represents N.
  • Z represents CH.
  • X and Z both represent CH. In another embodiment, X represents N and Z represents CH.
  • Y represents O.
  • R1, R2 and R3 are the same or different and are selected in the group comprising hydrogen atom, nitro, cyano, amino, alkyl, alkenyl, alkynyl, and alkoxy, preferably selected in the group comprising hydrogen atom, C 1-6 alkoxy and cyano.
  • R1 is H.
  • R1 represents H and R2 and R3 are the same or different and independently represent methoxy, - OCH 2 CH 2 OCH 3 , cyano, (1-aminocyclopropyl)methanolyl and (1-(dimethylamino)cyclopropyl)-methanolate.
  • R2 and R3 both represent an optionally substituted C 1-6 alkoxy group, preferably C 1-4 alkoxy group, more preferably methoxy.
  • R5, R6, R7, and R8 are the same or different and independently represent a hydrogen atom or a halogen atom. In a preferred embodiment, R5, R6, R7, and R8 are the same or different and independently represent a hydrogen, a chlorine or a fluorine atom.
  • At least one of R5, R6, R7, and R8 is a halogen atom, preferably chlorine or fluorine atom.
  • R8 represents a halogen atom, preferably chlorine or fluorine atom, more preferably a chlorine atom.
  • R5 represents H.
  • R6 represents H.
  • R7 represents H.
  • R5, R6 and R7 are hydrogen atoms.
  • R5, R6 and R7 are hydrogen atoms and R8 represents a halogen atom, preferably chlorine or fluorine atom, more preferably a chlorine atom.
  • R9 and R10 are the same or different and represent a hydrogen atom, alkyl, alkylcarbonyl, ONO 2 , OSO 2 R, wherein R is hydroxyl, alkyl, or cycloalkyl
  • R9 represents H.
  • R10 represents H.
  • R9 and R10 are both hydrogen atoms.
  • R11 is the group represented by the formula (i): wherein Q represent O, S or NH; preferably Q represent O, and R12 and R13 may be the same or different and represent hydrogen atom, halogen atom, alkyl, alkoxy, alkylthio, trifluoromethyl, nitro amino, alkoxycarbonyl, alkylcarbonyl, carbocyclic; preferably R12 and R13 may be the same or different and represent a hydrogen atom or a C 1-4 alkyl, more preferably a hydrogen atom or a methyl.
  • R11 is the group represented by the formula (ii): wherein Q represents O, S or NH, and R12 and R13 may be the same or different and represent a hydrogen atom, a halogen atom, alkyl, alkoxy, alkylthio, trifluoromethyl, nitro, amino, alkoxycarbonyl, alkylcarbonyl, or carbocyclic group; preferably R12 and R13 may be the same or different and represent hydrogen atom or a C 1-4 alkyl, more preferably a hydrogen atom or a methyl.
  • R11 is the group represented by the formula (iii): wherein Q represents O, S or NH; preferably Q represent S, and R12 and R13 may be the same or different and represent a hydrogen atom, a halogen atom, alkyl, alkoxy, alkylthio, trifluoromethyl, nitro, amino, alkoxycarbonyl, alkylcarbonyl, or carbocyclic; preferably R12 and R13 may be the same or different and represent a hydrogen atom; a chlorine atom; a bromine atom; methyl, t-butyl, acetyl or - CH 2 -COOEt.
  • R11 is the group represented by the formula (iv): wherein Q represents O, S or NH; preferably Q represent S, and R12 represents a hydrogen atom, a halogen atom, alkyl, alkoxy, alkylthio, trifluoromethyl, nitro, amino or carbocyclic group ; preferably R12 represents methyl, ethyl, t-butyl, trifluoromethyl, ethylthio, or cyclopropyl.
  • R13 is a hydrogen atom in groups (i) and (ii).
  • R11 represents an azolyl, alkyl, amido, carbocyclic or heterocyclic groups, preferably an azolyl of formula (i), (ii), (iii) or (iv), C 3-10 carbocyclic, C 3-10 heterocyclic, amide or C 1-6 alkyl.
  • R11 is selected from n- propyl, 2-MeOPh, 4-MeOPh, 2-MePh, 1-naphthyl, 5-chloropyridin-2-yl, 5-bromopyridin-2-yl, 5-methylpyridin-2-yl, 2,5-dichlorophenyl, 2-chloro-5-bromophenyl, 2-fluoro-5-chlorophenyl, 2-methoxy-5-chlorophenyl, 2-methoxy-5-bromophenyl, 2-methyl-5-fluorophenyl, 2-(difluoromethoxy)-5-chlorophenyl, or 5-methylisoxazo-3-yle.
  • R11 represents n -propyl or isoxazole group, preferably 5-methylisoxazo-3-yle.
  • compound for use of the invention is compound of formula (Ia) or a pharmaceutically acceptable salt or solvate thereof, wherein compound of formula (Ia) is represented hereunder: wherein R2, R3, R5, R6, R7, R8, R9, R10, R11, and X are as defined for compound of general formula I.
  • the compound of formula (Ia) is such that:
  • X represents CH. In another embodiment, X represents N.
  • R1 represents H and R2 and R3 are the same or different and independently represent methoxy, -OCH 2 CH 2 OCH 3 , cyano, (1-aminocyclopropyl)methanolyl and (1-(dimethylamino)cyclopropyl)-methanolate.
  • R2 and R3 both represent an optionally substituted C 1-6 alkoxy group, preferably C 1-4 alkoxy group, more preferably methoxy.
  • R2 and R3 are the same or different and are selected in the group comprising hydrogen atom, nitro, cyano, amino, alkyl, alkenyl, alkynyl, and alkoxy, preferably selected in the group comprising hydrogen atom, C 1-6 alkoxy and cyano.
  • R2 and R3 are the same or different and independently represent methoxy, -OCH 2 CH 2 OCH 3 , cyano, (1-aminocyclopropyl)methanolyl and (1-(dimethylamino)cyclopropyl)-methanolate.
  • R2 and R3 both represent an optionally substituted C 1-6 alkoxy group, preferably C 1-4 alkoxy group, more preferably methoxy.
  • R5, R6, R7, and R8 are the same or different and independently represent a hydrogen atom or a halogen atom. In a preferred embodiment, R5, R6, R7, and R8 are the same or different and independently represent a hydrogen, a chlorine or a fluorine atom.
  • At least one of R5, R6, R7, and R8 is a halogen atom, preferably chlorine or fluorine atom.
  • R6 represents H.
  • R7 represents H.
  • R5, R6 and R7 are hydrogen atoms.
  • R8 represents a halogen atom, preferably chlorine or fluorine atom, more preferably a chlorine atom.
  • R9 and R10 are the same or different and represent a hydrogen atom, alkyl, alkylcarbonyl, ONO 2 , OSO 2 R, wherein R is hydroxyl, alkyl, or cycloalkyl, preferably R9 and R10 are both hydrogen atoms.
  • R11 is an optionally substituted azolyl, preferably of formula (i), (ii), (iii) or (iv) as presented above.
  • R11 represents an azolyl, alkyl, amido, carbocyclic or heterocyclic groups, preferably an azolyl of formula (i), (ii), (iii) or (iv), C 3-10 carbocyclic, C 3-10 heterocyclic, amide or C 1-6 alkyl.
  • R11 is selected from n- propyl, 2-MeOPh, 4-MeOPh, 2-MePh, 1-naphthyl, 5-chloropyridin-2-yl, 5-bromopyridin-2-yl, 5-methylpyridin-2-yl, 2,5-dichlorophenyl, 2-chloro-5-bromophenyl, 2-fluoro-5-chlorophenyl, 2-methoxy-5-chlorophenyl, 2-methoxy-5-bromophenyl, 2-methyl-5-fluorophenyl, 2-(difluoromethoxy)-5-chlorophenyl, or 5-methylisoxazo-3-yle.
  • R11 represents n -propyl or isoxazole group, preferably 5-methylisoxazo-3-yle.
  • compound (I) is N- ⁇ 2-chloro-4-[(6,7-dimethoxy-4-quinolyl)oxy]-phenyl ⁇ -N'-(5-methyl-3-isoxazolyl)urea, also commonly named tizovanib (Av-951, KRN951), and represented by the following formula:
  • compound (I) is 1-(2-chloro-4-((6,7-dimethoxyquinazolin-4-yl)oxy)phenyl)-3-propylurea, and represented by the following formula:
  • the compound for use of the present invention may be used under the form of pharmaceutically acceptable salts thereof.
  • Preferred examples of such salts include: alkali metal or alkaline earth metal salts such as sodium salts, potassium salts or calcium salts; hydrohalogenic acid salts such as hydrofluoride salts, hydrochloride salts, hydrobromide salts, or hydroiodide salts; inorganic acid salts such as nitric acid salts, perchloric acid salts, sulfuric acid salts, or phosphoric acid salts; lower alkylsulfonic acid salts such as methanesulfonic acid salts, trifluoromethanesulfonic acid salts, or ethanesulfonic acid salts; arylsulfonic acid salts such as benzenesulfonic acid salts or p-toluenesulfonic acid salts; organic acid salts such as fumaric acid salts, succinic acid salts, citric acid salts, tart
  • Compounds of the present invention are for use in the prevention and/or treatment of diseases involving an activation of PDE4.
  • said diseases involving an activation of PDE4 may be neurological diseases, neurodegenerative diseases, inflammatory diseases, respiratory diseases, allergies, cardiovascular diseases, thrombosis, diabetes, cancer, rheumatoid arthritis, atopic dermatitis, psoriasis, asthma, inflammatory bowel disease.
  • the disease involving an activation of PDE4 is a neurodegenerative disease.
  • the neurodegenerative disease is Parkinson disease, Huntington disease, Alzheimer disease, multiple sclerosis, Amyotrophic Lateral Sclerosis, Spinal Muscular Atrophy or any disease wherein a progressive loss of structure or function of neurons, including death of neurons, occurs.
  • the neurodegenerative disease is Alzheimer's disease.
  • the disease involving an activation of PDE4 is cancer.
  • the cancer is a brain cancer, lung cancer, leukemia for example chronic lymphocytic leukaemia, osteosarcoma, and melanoma.
  • a therapeutically effective amount of compound for use of the invention may be administered prior to the onset of a disease, disorder, or condition involving an activation of PDE4. In an embodiment, a therapeutically effective amount of compound for use of the invention may be administered prior to the onset of a disease, disorder, or condition related to neurodegenerative disease, especially Alzheimer disease, for a prophylactic or preventive action.
  • the therapeutically effective amount of compound of the invention may be administered after initiation of a disease, disorder, or condition involving an activation of PDE4, preferably a disease, disorder, or condition related to neurodegenerative disease, especially Alzheimer disease, for a therapeutic action.
  • the therapeutically effective amount of compound of the invention may be administered prior to and after initiation of a disease, disorder, or condition involving an activation of PDE4, preferably a disease, disorder, or condition related to neurodegenerative disease, especially Alzheimer disease, for a prophylactic, preventive and/or therapeutic action.
  • the compounds for use of the present invention have an in vitro activity against PDE4.
  • tivozanib has an in vitro activity against PDE4.
  • a therapeutically effective amount of compound of the invention is an amount that is effective in reducing at least one symptom of at least one neurodegenerative disease, especially Alzheimer disease.
  • the therapeutically effective amount of compound may be appropriately determined in consideration of, for example, the age, weight, sex, difference in diseases, and severity of the condition of individual patients. It will be understood, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • the present invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the compound for use according to the present invention, or salt or solvate thereof, in association with a pharmaceutically acceptable vehicle.
  • the pharmaceutical composition of the invention is for use in the prevention and/or treatment of a disease selected from the group consisting of a neurodegenerative disease and a disease involving an activation of PDE4.
  • the compound for use of the invention or the pharmaceutical composition of the invention is administered to human or non-human animals.
  • the compound for use of the invention or the pharmaceutical composition of the invention is administered orally.
  • oral preparations include tablets, capsules, powders, granules, and syrups.
  • the compound for use of the invention or the pharmaceutical composition of the invention is administered parenterally, for example by intravenous injection, intramuscular injection, subcutaneous injection, intradermic injection, intraperitoneal injection, intracerebroventrocular (ICV) infusion, intracisternal injection or infusion.
  • parenterally for example by intravenous injection, intramuscular injection, subcutaneous injection, intradermic injection, intraperitoneal injection, intracerebroventrocular (ICV) infusion, intracisternal injection or infusion.
  • the compound for use of the invention or the pharmaceutical compositions of the invention is administered by inhalation spray, nasal, vaginal, rectal, sublingual, percutaneous or topical routes of administration.
  • the compounds for use of the invention may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
  • these various preparations may be prepared by conventional methods, for example, with commonly used pharmaceutically acceptable carriers, such as excipients, disintegrants, binders, lubricants, colorants, and diluents.
  • pharmaceutically acceptable carriers such as excipients, disintegrants, binders, lubricants, colorants, and diluents.
  • excipients include, for example lactose, glucose, corn starch, sorbit, and crystalline cellulose.
  • disintegrants include, for example starch, sodium alginate, gelatin powder, calcium carbonate, calcium citrate, and dextrin.
  • binders include, for example dimethylcellulose, polyvinyl alcohol, polyvinyl ether, methylcellulose, ethylcellulose, gum arabic, gelatin, hydroxypropylcellulose, and polyvinyl pyrrolidone.
  • lubricants include, for example talc, magnesium stearate, polyethylene glycol, and hydrogenated vegetable oils.
  • the pharmaceutical composition of the invention may further contain preparing buffers, pH adjusters, stabilizers, tonicity agents, and/or preservatives.
  • the content of the compound according to the invention in the pharmaceutical composition according to the invention may vary depending on the dosage form.
  • the compound according to the present invention may be administered without any other active ingredients which are usually applied in the prevention and/or treatment of the above mentioned pathological conditions.
  • the pharmaceutical composition of the present invention may further comprise other therapeutically active compounds which are usually applied in the prevention and/or treatment of the above mentioned pathological conditions.
  • the pharmaceutical compositions for the administration of the compounds for use of this invention may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients.
  • the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • compositions of the invention is in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions
  • the pharmaceutical compositions are in the form of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the compounds of the present invention are administered in the form of suppositories for rectal administration of the drug.
  • suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • creams, ointments, jellies, solutions or suspensions, tapes etc., containing the compounds of the present invention may be employed.
  • the invention further relates to a method of prevention and/or treatment of a subject suffering from a neurodegenerative disease and/or a disease involving an activation of phosphodiesterase-4, said method comprising administering to that patient a therapeutic effective amount of a compound as described in this invention or a pharmaceutically acceptable salt thereof.
  • a compound as described in this invention or a pharmaceutically acceptable salt thereof.
  • the compound is of formula (I) or (Ia). More preferably, the compound is tivozanib.
  • compounds of the invention may be used as psycho-stimulants, analgesics, antipyretics, psycholeptics or anxiolitics in the prevention and/or treatment of diseases involving an activation of PDE4.
  • PDE activities are measured by a two-step radioenzymatic assay as previously described at a substrate concentration of 1 ⁇ M cAMP or cGMP in the presence of 15,000 cpm [3H]-cAMP or [3H]-cGMP as a tracer, respectively.
  • a substrate concentration of 1 ⁇ M cAMP or cGMP in the presence of 15,000 cpm [3H]-cAMP or [3H]-cGMP as a tracer, respectively.
  • cyclic nucleotides are hydrolyzed into their respective nucleotide monophosphates.
  • This incubation is stopped by the addition of an excess of cAMP and cGMP in the presence of EDTA and a nonselective PDE inhibitor (theophylline) that inactivates PDE activity.
  • the second incubation performed in the presence of an excess of 5'-nucleotidase results in the formation of adenosine or guanosine.
  • This second incubation is stopped by the addition of an excess of adenosine and guanosine in the presence of EDTA.
  • the enzymatic reaction products are separated by anion-exchange column chromatography on QAE-Sephadex A-25 formate. The amount of resulting [3H]-nucleoside is determined by liquid scintillation.
  • [3H]-cAMP and [3H]-cGMP are purified by TLC on Silicagel 60 F254 TLC plates.
  • anion-exchange resin to recover [3H]-adenosine or [3H]-guanosine is critical for the accuracy of the assay. This method allows recovering and regenerating the resin, which can then be used over a long period of time. Because QAE-Sephadex A-25 is expensive, the ability to recycle is quite valuable.
  • Used resin collected from the assay columns is stored at +4°C and regenerated in a large glass column (35 ⁇ 8 cm) with a sintered-glass bottom by successive treatments:
  • This procedure allows separation of the dephosphorylated products (adenosine or guanosine) from the nonhydrolyzed cyclic nucleotide.
  • test tubes without PDE extract 50 ⁇ L of Tris-BSA
  • Tris-BSA 50 ⁇ L of Tris-BSA
  • the two first tubes, used to determine the total radioactivity (Rt) are directly poured into scintillation vials and 1.5 mL of formate is added; the two other tubes, used to determine blank assay (Ro), are treated similarly to the other test tubes.
  • CNP cyclic nucleotide phosphate
  • the inhibition concentration value (IC50) of tivozanib is of 4.8 ⁇ M in this assay.
  • the results are presented in figure 4 .
  • Cortical neurons from embryonic day 16-17 Wistar rat fetuses were prepared as follows. Dissociated cortical cells were plated at 4.5-5.0 10 4 cells/cm 2 in plastic dishes precoated with 1.5 mg/mL polyornithine (Sigma). Cells were cultured in a chemically defined Dulbecco's modified eagle's/F12 medium free of serum (Gibco) and supplemented with hormones, proteins and salts. Cultures were kept at 35°C in a humidified 5% CO 2 atmosphere, and at 6-7 DIV, cortical population was determined to be at least 97% neuronal by immunostaining.
  • a ⁇ low-n ⁇ -amyloid
  • the peptide used is A ⁇ 1-42 from Bachem (ref H1368, batch number: 1025459).
  • the oligomeric preparation contains a mixture of stable dimers, trimers and tetramers of A ⁇ 1-42 peptide, as well as monomeric forms of the peptide.
  • oligomers The same preparation of oligomers was used for all experimental set-ups and has been previously characterized in terms of oligomer composition (see figure 5 ), neurotoxicity in vitro as well as induction of cognitive impairment in mice.
  • tivozanib solution Tivozanib was dissolved in DMSO. A stock solution (10 mM DMSO) was prepared, and aliquots were stored at -20 °C before use.
  • HEK293 cells were cultured in DMEM medium containing 10 % FCS. Cells were plated in 12-well plates at a density of 400.000 cells per well and cultured for 24h at 37°C in a humidified 6% CO 2 atmosphere. Cells were then incubated for 1 h with increasing concentrations of tivozanib and then treated with vehicle or 25 ⁇ M forskolin for 15 minutes. Following washing steps and cell lysis, cellular level of cAMP level was monitored using an ELISA assay (Cyclic AMP XP assay kit - Cell signaling). Data are from 2 experiments performed in duplicate. Data are represented as fold increase of cAMP as compared to experiment using forskolin only, designed as 1.0 ( figure 7 ).
  • tivozanib is able to inhibit cAMP hydrolysis following activation of adenylate cyclase by forskolin.
  • This experiment shows that tivozanib interferes with cAMP metabolism through the modulation of PDE activity, most specifically PDE4-associated hydrolysis of cAMP.
  • tivozanib prevents from soluble low-n oligomer-induced neuronal cell death. Moreover, tivozanib is able to improve cellular cAMP level in living cells, showing that cellular signaling pathway(s) induced by tivozanib modulates the activity of PDE4.
  • Example 3 In vivo activity of tivozanib against PDE4
  • mice All animal experiments were carried out according to the National Institute of Health (NIH) guidelines for the care and use of laboratory animals, and approved by the French Ministry for Research and Technology. Animals were housed at a standard temperature (22 ⁇ 1°C) and in a light-controlled environment (lights on from 7 am to 8 pm) with ad libitum access to food and water. Upon arrival, Male C57BL/6 mice (C57BL/6J Rj, ref SC-CJ-12w-M, Janvier, France) were group housed. From 1 week before the experiment started and until the end of the experiment, mice were individually housed. Animals were monitored twice-a-day by laboratory personnel (8 am and 4 pm). In case general health status of an animal is significantly worsens, the mouse was sacrificed. Definitions of acceptable endpoints are as follow: no spontaneous movements and inability to drink or eat in 24-h observation period, massive bleeding, spontaneous inflammation (general observations of mouse skin and eyes), or missing anatomy.
  • NASH National Institute of Health
  • soluble A ⁇ oligomers 50 pmol in 1 ml or vehicle (saline, 1 ml) were injected into the right ventricle.
  • vehicle saline, 1 ml
  • the stereotaxic coordinates from the bregma were as follow (in mm): AP-0.22, L-1.0 and D-2.5. Injections were made using a 10 ml Hamilton microsyringe fitted with a 26-gauge needle.
  • the series of arm entries were recorded manually and the arm entry was considered to be completed when the hind paws of the mouse are completely placed in the arm. Alternation is defined as successive entries into the 3 arms on overlapping triplet sets. The percentage of alternation was calculated as the ratio of actual (total alternations) to possible alternations (defined as the number of arm entries minus 2), multiplied by 100.
  • Formulation of tivozanib and animal treatment In order to administrate tivozanib through per os gavage, a 3 mg/mL tivozanib solution was prepared in 0.5% (w/v) methylcellulose. The solution was stirred (magnetic stirring) overnight protected from light. Animals were feed with tivozanib at a dose of 30 mg/kg/day from day -2 to day +4. At day 0, soluble A ⁇ low-n oligomers were icv administrated (induction of the disease) and spatial working memory performances measured at day +4 using the Y-maze test.
  • mice per group are defined as follows:
  • Spontaneous alternation behavior which is considered to be a measure of immediate (short-term) spatial working memory performance, was investigated using the Y-maze test performed 4 days after icv injection of saline or A ⁇ oligomers.
  • Spontaneous alternation (A) and the number of arm entries (B) were measured during a 5 min session ( figure 8 ).
  • Data are presented as mean ⁇ S.E.M. p ⁇ 0.05 vs. A ⁇ -treated mice.
  • mice receiving a vehicle gavage and icv injected with 50 pmol A ⁇ 1-42 oligomers displayed significantly impaired spatial working memory (p ⁇ 0.05 as compared to mice receiving a vehicle gavage and icv injected with vehicle) ( figure 8A ).
  • mice receiving tivozanib by gavage and icv injected with A ⁇ oligomers are not statistically different from control group A (vehicle gavage and vehicle icv injection) ( figure 8A ).
  • animals from group C displayed a normal behavior and their performances are not statistically different from groups A and D.
EP12181152.5A 2012-02-13 2012-08-21 Verbindung zur Verwendung zur Prävention bzw. Behandlung einer neurogenerativen Erkrankung oder einer Erkrankung mit Phosphodiesterase (PDE4) -4-Aktivierung Withdrawn EP2626073A1 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/EP2013/052867 WO2013120896A1 (en) 2012-02-13 2013-02-13 Compound for use in the prevention and/or treatment of a neurogenerative disease or a disease involving an activation of phosphodiesterase-4 (pde4)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US201261598139P 2012-02-13 2012-02-13

Publications (1)

Publication Number Publication Date
EP2626073A1 true EP2626073A1 (de) 2013-08-14

Family

ID=46704534

Family Applications (1)

Application Number Title Priority Date Filing Date
EP12181152.5A Withdrawn EP2626073A1 (de) 2012-02-13 2012-08-21 Verbindung zur Verwendung zur Prävention bzw. Behandlung einer neurogenerativen Erkrankung oder einer Erkrankung mit Phosphodiesterase (PDE4) -4-Aktivierung

Country Status (2)

Country Link
EP (1) EP2626073A1 (de)
WO (1) WO2013120896A1 (de)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016114322A1 (ja) * 2015-01-13 2016-07-21 国立大学法人京都大学 筋萎縮性側索硬化症の予防及び/又は治療剤

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0860433A1 (de) 1995-11-07 1998-08-26 Kirin Beer Kabushiki Kaisha Chinolinderivate und chinazolinderivate welche die autophosphorylierung des von blutplättchen abstammenden wachstumsfaktorrezeptors inhibiren und sie enthaltende pharmazeutische zusammensetzungen
EP1153920A1 (de) 1999-01-22 2001-11-14 Kirin Beer Kabushiki Kaisha Chinolinderivate und chinazolinderivate
EP1382604A1 (de) 2001-04-27 2004-01-21 Kirin Beer Kabushiki Kaisha Chinolinderivate mit einer azolylgruppe und chinazolinderivate
EP1447405A1 (de) 2001-10-17 2004-08-18 Kirin Beer Kabushiki Kaisha Chinolin- oder chinazolinderivate, die die autophosphorylierung von rezeptoren für den fibroblastenwachstumsfaktor hemmen
EP1535910A1 (de) 2002-05-01 2005-06-01 Kirin Beer Kabushiki Kaisha Chinolinderivate und chinazolinderivate, die die autophosphorilierung des macrophage colony stimulating factor receptors inhibieren
EP1566379A1 (de) 2002-10-29 2005-08-24 Kirin Beer Kabushiki Kaisha CHINOLINDERIVATE UND CHINAZOLINDERIVATE ALS HEMMER DER Flt3-AUTOPHOSPHORYLIERUNG UND DIESE ENTHALTENDE MEDIZINISCHE ZUSAMMENSETZUNGEN
WO2007136225A1 (en) * 2006-05-24 2007-11-29 Industry-Academic Cooperation Foundation, Yonsei University A method for inhibiting angiogenesis using dkk1 and composition comprising the same

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2010010172A (es) * 2008-03-17 2010-11-25 Ambit Biosciences Corp Derivados de quinazolina como moduladores de quinasa raf y metodos de uso de los mismos.
WO2011046991A2 (en) * 2009-10-15 2011-04-21 Gilead Connecticut, Inc. Certain substituted ureas as modulators of kinase activity

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0860433A1 (de) 1995-11-07 1998-08-26 Kirin Beer Kabushiki Kaisha Chinolinderivate und chinazolinderivate welche die autophosphorylierung des von blutplättchen abstammenden wachstumsfaktorrezeptors inhibiren und sie enthaltende pharmazeutische zusammensetzungen
EP1153920A1 (de) 1999-01-22 2001-11-14 Kirin Beer Kabushiki Kaisha Chinolinderivate und chinazolinderivate
EP1384712A1 (de) 1999-01-22 2004-01-28 Kirin Beer Kabushiki Kaisha Derivate des N-((Chinolinyl)oxy)-phenyl)-Harnstoffs und des N-((Chinazolinyl)oxy)-phenyl)-Harnstoffs mit Antitumor Aktivität
EP1382604A1 (de) 2001-04-27 2004-01-21 Kirin Beer Kabushiki Kaisha Chinolinderivate mit einer azolylgruppe und chinazolinderivate
EP1652847A1 (de) 2001-04-27 2006-05-03 Kirin Beer Kabushiki Kaisha Chinolin- und Chianzolinderivate zur Behandlung von Tumoren
EP1447405A1 (de) 2001-10-17 2004-08-18 Kirin Beer Kabushiki Kaisha Chinolin- oder chinazolinderivate, die die autophosphorylierung von rezeptoren für den fibroblastenwachstumsfaktor hemmen
EP1535910A1 (de) 2002-05-01 2005-06-01 Kirin Beer Kabushiki Kaisha Chinolinderivate und chinazolinderivate, die die autophosphorilierung des macrophage colony stimulating factor receptors inhibieren
EP1566379A1 (de) 2002-10-29 2005-08-24 Kirin Beer Kabushiki Kaisha CHINOLINDERIVATE UND CHINAZOLINDERIVATE ALS HEMMER DER Flt3-AUTOPHOSPHORYLIERUNG UND DIESE ENTHALTENDE MEDIZINISCHE ZUSAMMENSETZUNGEN
WO2007136225A1 (en) * 2006-05-24 2007-11-29 Industry-Academic Cooperation Foundation, Yonsei University A method for inhibiting angiogenesis using dkk1 and composition comprising the same

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BURGIN ET AL., NATURE BIOTECHNOLOGY, vol. 28, no. 1, January 2010 (2010-01-01), pages 63 - 72
DE FILIPPIS DANIELE ET AL: "Are anti-angiogenic drugs useful in neurodegenerative disorders?", CNS & NEUROLOGICAL DISORDERS, BENTHAM SCIENCE PUBLISHERS LTD, NL, vol. 9, no. 6, 1 December 2010 (2010-12-01), pages 807 - 812, XP009164397, ISSN: 1871-5273 *
DESBENE ET AL., NEUROBIOLOGY OF AGING, vol. 33, 2012, pages 1123.E17 - 1123.E,29
ESKENS FERRY A L M ET AL: "Biologic and clinical activity of tivozanib (AV-951, KRN-951), a selective inhibitor of VEGF receptor-1, -2, and -3 tyrosine kinases, in a 4-week-on, 2-week-off schedule in patients with advanced solid tumors.", CLINICAL CANCER RESEARCH : AN OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH 15 NOV 2011 LNKD- PUBMED:21976547, vol. 17, no. 22, 15 November 2011 (2011-11-15), pages 7156 - 7163, XP002686635, ISSN: 1078-0432 *
GARCIA ET AL., THE JOURNAL OF NEUROSCIENCE, vol. 30, no. 22, 2 June 2010 (2010-06-02), pages 7516 - 7527
PAGÈS LLUÍS ET AL: "PDE4 inhibitors: a review of current developments (2005 - 2009).", EXPERT OPINION ON THERAPEUTIC PATENTS NOV 2009 LNKD- PUBMED:19832118, vol. 19, no. 11, November 2009 (2009-11-01), pages 1501 - 1519, XP002686636, ISSN: 1744-7674 *
UEMURA YASUNORI ET AL: "The selective M-CSF receptor tyrosine kinase inhibitor Ki20227 suppresses experimental autoimmune encephalomyelitis.", JOURNAL OF NEUROIMMUNOLOGY MAR 2008 LNKD- PUBMED:18378004, vol. 195, no. 1-2, March 2008 (2008-03-01), pages 73 - 80, XP002686634, ISSN: 0165-5728 *
VAGNUCCI A H ET AL: "Alzheimer's disease and angiogenesis", THE LANCET, LANCET LIMITED. LONDON, GB, vol. 361, no. 9357, 15 February 2003 (2003-02-15), pages 605 - 608, XP004778573, ISSN: 0140-6736, DOI: 10.1016/S0140-6736(03)12521-4 *
YOUSSEF ET AL., NEUROBIOLOGY OF AGING, vol. 29, 2008, pages 1319 - 1333

Also Published As

Publication number Publication date
WO2013120896A1 (en) 2013-08-22

Similar Documents

Publication Publication Date Title
US11530219B2 (en) Ligands to cereblon (CRBN)
US10112922B2 (en) Inhibitor of bruton's tyrosine kinase
JP6167173B2 (ja) T細胞受容体を調節することができる複素環およびそれを使用するための方法
US7550465B2 (en) Pyrido[3,2-e]pyrazines, their use as inhibitors of phosphodiesterase 10, and processes for preparing them
US8518958B2 (en) Pyrido [2,3-D] pyrimidines and their use as kinase inhibitors
CN108349964A (zh) N-(吡啶-2-基)-4-(噻唑-5-基)嘧啶-2-胺类化合物作为治疗性化合物
EP3810608A1 (de) Oga-inhibitor-verbindungen
EP3697781B1 (de) Antagonisten des muskarinischen acetylcholinrezeptors m4
KR20160076654A (ko) 엑소솜을 유효성분으로 함유하는 세포 노화 예방 또는 치료용 약학조성물
US20140163006A1 (en) Pyrido-[2,3-D] Pyrimidines and Their Use as Kinase Inhibitors
US20220402938A1 (en) Glutamate transporter activators and methods using same
WO2018053373A1 (en) Uses of satl-inducible kinase (sik) inhibitors for treating osteoporosis
MX2014014657A (es) Heterociclos bicíclicos capaces de modular las respuestas de los linfocitos t, y métodos de uso de los mismos.
US20210269442A1 (en) Fused ring derivative used as fgfr4 inhibitor
EP3724194B1 (de) Substituierte azetidin-dihydrothienopyrimidine und deren verwendung als phosphodiesterase-inhibitoren
KR20240017910A (ko) 인지 향상제로서 line-1 억제제
EP2626073A1 (de) Verbindung zur Verwendung zur Prävention bzw. Behandlung einer neurogenerativen Erkrankung oder einer Erkrankung mit Phosphodiesterase (PDE4) -4-Aktivierung
TW201331202A (zh) [1,2,4]三唑并吡啶及其作為磷酸二酯酶抑制劑之用途
WO2009126335A2 (en) Ant2 inhibitor compounds and methods of use thereof
US20230241025A1 (en) Antiviral use of fabp4 modulating compounds
JP2024518143A (ja) ニトロフェニル-アクリルアミドおよびその使用
WO2024073283A1 (en) Heterocycles as modulators of nsd activity
EP4288441A1 (de) Verfahren zur behandlung von spinocerebellarer ataxie typ 3
WO2022109292A1 (en) Methods and materials for inhibiting cb1 activity
WO2019046534A1 (en) INDOLOQUINOLINE COMPOUNDS DEUTTERED

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

17P Request for examination filed

Effective date: 20140214

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20140215